You are on page 1of 10

Eur J Haematol 2004: 72: 79–88 Copyright  Blackwell Munksgaard 2004

Printed in UK. All rights reserved


EUROPEAN
JOURNAL OF HAEMATOLOGY

Review article

Monoclonal antibodies in the treatment


of autoimmune cytopenias
Robak T. Monoclonal antibodies in the treatment of autoimmune Tadeusz Robak
cytopenias. Department of Hematology, Medical University of
Eur J Haematol 2004: 72: 79–88.  Blackwell Munksgaard 2004. Lodz and Copernicus Hospital, Lodz, Poland

Abstract: In recent years, clinical studies have been undertaken with


selected monoclonal antibodies (MoAbs) in the treatment of several
hematological diseases, especially in malignant disorders. However,
some clinical observations indicate that MoAbs may be an important
alternative for the conventional therapy of some autoimmune disorders.
Two MoAbs directed against CD20 antigen (rituximab, Rituxan,
Mabthera) and CD52 antigen (alemtuzumab, Campath-1H) seem to be
especially useful for this purpose. Autoimmune cytopenias have been
investigated in the last few years with positive preliminary results.
Key words: rituximab; alemtuzumab; campath-1H;
Rituximab seems to be an effective and safe agent for the treatment of autoimmune hemolytic anemia; immune
immune thrombocytopenias, autoimmune hemolytic anemia, cold thrombocytopenia; cold agglutinin disease; pure red cell
agglutinin disease and pure red cell aplasia. Although the case series are aplasia
small, rituximab seems to be an effective and safe agent for the treatment
Correspondence: Tadeusz Robak, Department
of these diseases. Clinical experience with alemtuzumab in patients with of Hematology, Medical University of Lodz, 93-513
autoimmune cytopenias is even more limited than with rituximab. Lodzd, Pabianicka 62, Poland
However, preliminary results indicate that further studies with this Tel: +4842 6895191
MoAb are warranted. A longer follow-up and the studies on larger Fax: +4842 6895192
number of patients are needed to determine the real value of these new e-mail: robaktad@onet.pl
approaches in autoimmune cytopenias. Recent experiences with the use
of MoAbs in treatment of these diseases are the subject of this review. Accepted for publication 23 October 2003

The autoimmune cytopenias (AC) include auto- in other patients specific therapy for cytopenia is
immune hemolytic anemia (AIHA), pure red cell needed. The majority of patients with AC require
aplasia (PRCA), immune thrombocytopenia (ITP), only standard immunosuppression, mainly steroids,
autoimmune neutropenia (AIN) and various com- to achieve long-lasting remission. In a proportion
binations of these disorders, such as autoimmune of refractory disease, second-line treatment such as
pancytopenia and EvansÕ syndrome (1). These splenectomy, or cytotoxic drugs for ITP or AIHA
disorders are idiopathic or associated with other and granulocyte-colony stimulating factor (G-CSF)
malignant or non-malignant diseases including for AIN is needed. However, as some patients are
lymphoma, chronic lymphocytic leukemia (CLL), resistant to conventional therapy or have significant
systemic lupus erythematosus (SLE), rheumatoid side effects after administration of immunosuppres-
arthritis, Sjogren’s disease and ulcerative colitis (2). sive agents, they need an alternative treatment.
Despite the complex pathogenetic mechanism of In the last few years monoclonal antibodies
autoimmune cytopenias, B cells play a crucial role (MoAbs) directed against B and/or T cells have
in the pathogenesis of these disorders (3). They are become a new approach in treating patients with
responsible for the production of autoantibodies severe, refractory AC, who have chronic refractory
that are directly or indirectly (e.g. immune complex or relapsing cytopenias and suffer life-threatening
formation) destructive. These cells also act as anemia, hemorrhages or infections (1–7).
highly efficient antigen presenting cells supporting
the activation and auto-reactivity of T cells
Monoclonal antibodies
involved in the process (3). The treatment of
associated neoplastic or autoimmune diseases may The most important clinical value in the patients
induce remission of AC in some patients. However, with autoimmune cytopenias has human-mouse

79
Robak

antibody rituximab (Rituxan, Mabthera, F. ies which can have an impact on the responsive-
Hoffman-La Roche Ltd, Basel, Switzerland) that ness. These host responses could attenuate
targets CD20 antigen on B lymphocytes (5–7, 8). response to re-treatments and potentially mediate
Preliminary reports indicate that alemtuzumab allergic reactions.
(Campath-1H, JLEX Pharmaceuticals, San Anto- Alemtuzumab is a humanized rat IgG1 anti-
nio, TX, USA), a humanized rat immunoglobulin CD52 MoAb that binds to the cell membrane of
G1 (IgG1) anti-CD52 MoAb that binds to the cell virtually all normal as well as malignant lympho-
membrane of virtually all B and T-cells is also cytes, whether B cells or T cells. Similar to rituxim-
active in the treatment of AC (1, 4, 9, 10). ab, alemtuzumab has the ability to cause cell lysis
Rituximab was the first MoAb approved in 1997 using a host–effector mechanism such as comple-
by the Food and Drug Administration (FDA) for ment fixation, ADCC and induction of apoptosis
the treatment of cancer (11). This MoAb demon- (9, 10). Administration of alemtuzumab results in
strates significant activity in patients with various prolonged and profound peripheral blood lymp-
lymphoid malignancies, including indolent and hopenia particularly affecting T cells. Alemtuzumab
aggressive forms of B-cell non-Hodgkin’s lym- is highly active in cases of CLL are even refractory
phoma (NHL) and B-cell CLL (8, 10). Rituximab to fludarabine, and in the T-cell variant of pro-
rapidly eliminates most circulating B cells, suggest- lymphocytic leukemia (T-PLL). The drug doses
ing that it could be beneficial in autoantibody- should be increased gradually. In the first week the
mediated diseases by targeting the auto-reactive B first dose of 3 mg should be administered and then
cells (11). Moreover, some studies on other auto- increased to 10 mg and subsequently to 30 mg as
immune diseases did not show any correlation soon as infusion-related reactions are tolerated.
between the decline of autoantibody levels and In CLL, alemtuzumab is usually administered
response, suggesting that additional mechanisms three times per week for at least 12 wk intraven-
involving antigen presentation and help to T cells ously or subcutaneously. This MoAb has also been
are important (7). Complement-dependent cytotox- incorporated in novel conditioning regimens
icity, antibody-dependent cell-mediated cytotoxi- designed to facilitate stem cell transplantation in
city (ADCC) and induction of apoptosis indicate hematological malignancies. Similar to rituximab,
that rituximab is cytotoxic to CD20-positive cells. flu-like symptoms after the first doses of ale-
However, CD20 is not expressed on all B-cell mtuzumab, especially when administered intraven-
forms. In particular, progenitors, as well as plasma ously, are a frequent event. More importantly this
cells, may be devoid of CD20 and hence unrespon- agent induces immunosuppression which lasts for
sive to this agent. Thus, after rituximab treatment several months after the cessation of treatment,
plasma cells may still continue to be present – leading to increased susceptibility to infections. For
inducing autoantibodies for months or even years this reason anti-infective prophylaxis is mandatory.
(3). Moreover, it is unknown whether all CD20 B
cells are equally susceptible to rituximab-mediated
Autoimmune thrombocytopenia
deletion. Until now, there is little or no data
regarding depletion of B cells at sites other than Autoimmune thrombocytopenia [idiopathic thrombo-
blood. Moreover, there may be pro-survival factors cytopenic purpura (ITP)] is an autoimmune dis-
acting in certain autoimmune diseases, or in order characterized by persistent thrombocytopenia
lymphoproliferation-associated conditions, which because of autoantibody binding to platelet anti-
may protect B cells from this agent. gens causing their premature destruction by the
Rituximab is administered as an intravenous reticuloendothelial system (12). Traditionally, chro-
infusion with a recommended dosage of 375 mg/ nic ITP is defined as persistence of a platelet count
m2, once weekly for 4 wk. Treatment with this of <150 · 109/L for more than 6 months. In
agent is usually well tolerated. However, infusion- general, patients with platelet counts exceeding
related reactions occur in the majority of patients. 30 · 109/L require no treatment unless they are
These adverse events are typically fever, chills, undergoing a procedure likely to induce blood loss
rigors and rarely hypertension and bronchospasm, (13). First-line therapy comprises oral corticoster-
although the incidence of these side effects oids and high doses of intravenous Ig (HDIVIg).
decrease with subsequent rituximab infusion. Splenectomy is usually considered as a second-line
Moreover, the prolonged impairment of antibody therapy. In patients refractory to splenectomy
production increases the risk of viral and bacterial having significant thrombocytopenia and hem-
infections. It should also be remembered that orrhagic symptoms, other therapeutic modalities
rituximab is a human–mouse chimeric antibody, including immunosuppressive agents such as vincr-
and hence, treated patients may be susceptible to istine, azathioprine, cyclophosphamide, vinca alka-
the development of human anti-chimeric antibod- loids, danazol, plasmapheresis, dapsone, and

80
Monoclonal antibodies in autoimmune cytopenias

intravenous anti-D for Rh(D)-positive patients can

Responses only after higher doses

and responded for retreatment


be used. Those who failed to respond to standard

new response after relapse


first- and second-line therapy and who required

Two patterns of response:


In two patients R induced
Comments
treatment with monoclonal antibodies anti-CD20

Two patients relapsed


in younger patients
(rituximab) or alemtuzumab were evaluated in

early and late


Better response
several studies (14–23). The results of larger trials
are presented in Table 1.
Saleh et al. (18) reported the preliminary results
of a prospective pilot phase I/II clinical trial in
which they enrolled 13 patients with ITP who had
failed corticosteroid therapy and whose platelet

>6 m in seven patients


count was <75 · 109/L. Rituximab was adminis-

Response duration

6 m in two patients;
3 m in one patient
tered in a dose-escalating fashion using doses
ranging from 50 to 375 mg/m2, weekly for 4 wk.

60 € 480 d

21–455+ d

21–455+ d
7–56+ wk
Twelve patients completed treatment and were
evaluable for response. Rituximab-associated toxi-
city consisting of infusion-related fever and chills
was seen in less than one-third of patients and none
of the patients discontinued treatment because of

5 (20)

9 (45)

4 (57)

5 (41)

24 (31)
CR (%)

1 (8)
toxicity. None of the three patients who received
the lowest doses (50 mg/m2 followed by 150 mg/

Response
m2) achieved a clinical response. However, these
doses did not effectively deplete B cells. Three of

OR (%)

13 (52)

3 (25)

13 (65)

6 (86)

9 (75)

44 (58)
nine patients (30%) who have received rituximab at
doses close to or equal to the full dose had objective
clinical response including two complete response

R 50 to 375 mg/m2/weekly · 4
(CR) and one partial response (PR). All responding
patients have maintained their platelet count longer R 375 mg/m2/weekly · 4

R 375 mg/m2/weekly · 4

R 375 mg/m2/weekly · 4

R 375 mg/m2/weekly · 4
than 2 months without additional therapy.
Treatment
regimen

Subsequently Stasi et al. (14) presented the


results of a prospective study in 25 patients with

R, rituximab; pt, patients; d, day, wk, week; m, month; yr, year; OR, overall response; CR, complete response.
ITP treated with rituximab at a dose of 375 mg/m2
once weekly for 4 wk. The patients were resistant to
previous two–five different therapeutic options
including eight patients who had already failed
splenectomy. Five patients showed a CR (platelet
Table 1. Larger studies evaluating the efficacy of rituximab in immune thrombocytopenia

Refractory or relapsed

Refractory or relapsed
count >100 · 109/L) and a PR (platelet count
Refractory to steroids

Refractory to steroids
Refractory/relapsed
different regimens

between 50 and 100 · 109/L) was seen in another


characteristic

Refractory to 2–5
PatientsÕ

five. In additional three patients minor response


(platelet count below 50 · 1010/L), with no need for
continued treatment was observed giving an overall
response rate of 52%. In seven cases responses were
sustained for 6 months or longer. It is worth noting
that in some patients with relapsed disease repeated
46 (22–74)

40 (20–66)

challenge with rituximab induced a new response.


Age
(yr)

21–77

28–71

16–77

In responding patients a significant rise in platelet


>16

counts was observed usually 1 wk after the first


rituximab infusion. More recently, the same
authors reported the results of the treatment with
of patients
Number

rituximab in seven additional cases with ITP (16).


25

12

20

12

76

In this group the platelet count rose to >50 ·


109/L in six patients, including four patients
achieving CR and two PR. In contrast to the
Saleh et al. (18)
Stasi et al. (14)

Stasi et al. (16)

previous study, in five of the responders there was


Zaja et al. (19)

Giagounidis

no significant increase in the platelet count during


Reference

et al. (23)

rituximab treatment. Patients began to respond


Total

only 2–5 wk after last antibody administration with

81
Robak

peak platelet count occurring 10–16 wk after the evaluated patients responded to the treatement. In
start of treatment. three patients the remission lasted more than
Stasi et al. (14, 16) identified two patterns: early 4–9 months. However, worsening of thrombocy-
response with an increase in platelet count after the topenia during antibody therapy was observed in
first or second infusion and late response with rise two patients and in most cases the response began
of platelet count at weeks 6–8. It is possible that in between 4 and 6 wk after the start of the antibody
patients with early response, opsonized B cells therapy.
block the reticulo-endothelial system. In late More recently Willis et al. (4) described 21
responders the decreased production of antiplatelet patients with severe and life-threatening autoim-
antibodies is responsible for an increase in the mune cytopenias resistant to standard immunosup-
platelet count (16). However, no clear explanation pression therapies and treated with alemtuzumab.
for the effect of rituximab or for the lack of In this group one patient had ITP, three ITP and
response could be found in those and other studies AIHA (EvansÕ syndrome), three ITP, AIHA and
(14, 16, 19, 23). In a group of 20 patients with ITP AIN, one ITP and AIN and one ITP and PRCA.
treated with standard doses of rituximab the drug Alemtuzumab was administered at a dose of 10 mg/
proved to be active in 13 patients with nine d for 10 d. A response was seen in two patients with
achieving CR, (19). No correlation was observed EvansÕ syndrome but both relapsed in 3 months. A
between response and gender, time between diag- patient with ITP with severe hemorrhages associ-
nosis and treatment, total and CD20+ lymphocyte ated with AIN had a sustained response. One
count, level of CD20 expression on B cells before patient with ITP and PRCA responded to ITP but
the therapy and pharmacokinetics of the drug. relapsed 3 months later. It should be noted that
However, older age seems to be the factor corre- patients entering the study received cyclosporine
lated with a course response rate (5). after alemtuzumab in an attempt to reduce the
It should be noted that rituximab is active not incidence of relapse. Alemtuzumab was well toler-
only in ITP but also in secondary immunothromb- ated in all patients apart from first-day reactions,
ocytopenia in the patients with connective tissue observed in most patients. Alemtuzumab is cur-
diseases, chronic graft vs. host disease and B-cell rently approved for the treatment of B-CLL resist-
lymphoproliferative disorders, including those with ant to alkylating agents and fludarabine (10).
purine nucleoside treatment associated ITP (19, 24– However, in this disease the drug is administered
26). Hegde and his colleagues (25) reported three at a dose of 30 mg in 2 h i.v. infusion three times
CLL patients with refractory fludarabine-associ- weekly for a maximum period of 12 wk. It is
ated ITP treated with standard doses of rituximab possible, that the method of its administration in
(375 mg/m2/wk for 4 wk). The patients achieved a ITP and other cytopenias is not optimal and better
platelet count of >50 · 109/L on day 21. These results could be obtained with longer treatment.
results indicate that rituximab can rapidly reverse
refractory fludarabine-associated ITP. Similarly,
Autoimmune hemolytic anemia
Hensel and Ho (27) described a case of the hairy
cell leukemia with autoimmune thrombocytopenia Autoimmune hemolytic anemia results from the
after treatment with pentostatin. One week after the production of pathologic antibodies that bind to
first administration of rituximab, the peripheral red blood cells and lead to their destruction. These
blood count recovered with a normal platelet syndromes can be classified according to the
count. The mechanism by which purine nucleoside characteristic temperature activity of the antibodies
treatment-associated cytopenias respond to the (28). The established treatment of the warm type of
treatment with rituximab is not clear. However, AIHA consists of corticosteroids in monotherapy
some authors hypothesize that profound suppres- or combined with azathrioprine or cyclophospha-
sion of CD4 cells by fludarabine or cladribine may mide. In secondary AIHA, a search for an under-
cause aberrations of the immunoregulatory circuits lying factor that requires specific therapy, such as
involving malignant B cells and on this basis it is lymphoproliferative connective tissue disease, is
presumed that B-cell-mediated autoimmune com- indicated. In steroids refractory patients splenecto-
plications may improve by anti-CD20 therapy (25). my may be indicated.
There is not much literature on the use of Rituximab appeared to have efficacy in the
alemtuzumab in the treatment of ITP. However, treatment of AIHA, both idiopathic or associated
10 yr ago Lim et al. (21) first reported the results of with B-cell chronic lymphoproliferative disorders
treatment with this MoAb in six patients with ITP (Table 2) (7, 29–44). Although the case series is
refractory to conventional therapy. Three patients small, interesting results have been observed in
had underlying CLL/non-Hodgkin’s lymphoma idiopathic AIHA with warm autoantibodies in
and one had Hodgkin’s disease. Four of five children. Quartier and colleagues (29) treated five

82
Monoclonal antibodies in autoimmune cytopenias

children with severe idiopathic AIHA and one child

Infections complications in two patients


and 2nd response after retreatment
with autoimmune hemolysis after stem cell
transplantation with rituximab. All patients were

Long-lasting B-cell deficiency


refractory to glucocorticosteroids and two also to

Four patients in CR of LD
Comments

Three patients relapsed


splenectomy. Four rituximab infusions were given

All patients with CLL


at a standard dose of 375 mg/m2 once per week.
Two patients received eight additional infusions at
the same dose over 14 wk. All patients remained in
complete remission 15–22 months after the start of
rituximab therapy. Moreover, corticosteroids and
immunosuppressive drugs could be stopped or their
doses markedly reduced. It is important to note, that
M 13 m (7–23+)

R, rituximab; m, months; w, week; m, month; yr, year, M, median; Ig, immunoglobulins; NR, not reported; CLL, chronic lymphocytic leukemia; LD, lymphoproliferative disorders; pt, patients.
M 8 m (3–20)
there were no infusion-related side effects and severe
Response
duration

7.3–27.6 m

infections. These results have recently been con-


3–22 m
15–22 m

firmed by Zecca and colleagues (7) in a group of 15


NR

children treated with rituximab. This is the largest


series of children with AIHA treated with this agent
6 (100)
CR (%)

reported so far. In 13 cases, warm-reactive autoan-


3 (60)

9 (24)
NR

NR
NR

tibodies of the IgG type were demonstrated by the


Response

direct antiglobulin test. In four children a concom-


itant autoimmune disease was present at the time
6 (100)

4 (100)
8 (100)
5 (100)
OR (%)

13 (87)

36 (95)

of AIHA diagnosis. All patients had previously


received two or more courses of immunosuppressive
therapy. Most of the patients were given three
infusions of rituximab. With a median follow-up of
R 375 mg/m /weekly · 4–12 w
R 375 mg/m2/weekly · 2–4 w

R 375 mg/m2/weekly · 4–6 w

15 months 13 patients responded and two did not


R 375 mg/m2/weekly · 2–5
R 375 mg/m2/weekly · 4
Treatment regimen

respond. Three patients relapsed 7, 8 and 10 months


after completion the treatment, respectively, and all
three patients were re-treated with rituximab, achiev-
2

ing remission. Four further children were treated by


Motto et al. (38). Who received four to six doses of
rituximab at a dose of 375 mg/m2. All four patients
became transfusion independent and were taken off
prednisone completely.
Refractory to steroids and/or Ig

Some recent reports indicate that rituximab


PatientsÕ characteristic

should be considered in the management of lympho-


Table 2. Larger studies evaluating efficacy of rituximab in autoimmune hemolytic anemia

Refractory to prednisone

Refractory to prednisone

and chemotherapy all

proliferative disorders (LD) with steroid refractory


Refractory to steroids

Refractory to steroids

patients with LD

AIHA (33–37). Trape and colleagues (36) reported


five cases of LD who were treated successfully with
rituximab resulting in a clinical improvement of both
the AIHA and of the LD. Two patients had large
B-cell NHL, two had B-CLL and one had B-cell
prolymphocytic leukemia (PLL). All the patients
were initially treated with chemotherapy but none of
60 (46–70) yr
age (range)
Median

0.3–14 yr

0.3–70 yr
3.5–4 yr

them achieved a stable improvement of AIHA. The


44–66 yr
7–35 m

median time from chemotherapy to rituximab


administration was 3 months. The MoAb was
administered at standard doses of 375 mg/m2/wk
of patients

for 4 wk. All patients showed a recovery from AIHA


Number

6
15

4
8
5

38

and three of them also had CR. All three patients


were alive at first CR of AIHA and LD at the time of
publication from 8 to 20 months (median 8 months).
Iannitto and colleagues (37) used rituximab as rescue
Quartier et al. (29)

Motto et al. (38)


Gupta et al. (33)
Trape et al. (36)

therapy for a patient with CLL and secondary


Zecca et al. (7)

AIHA. He was previously treated with chlorambucil


Reference

and prednisone, fludarabine, mitoxantrone, and


Total

dexamethasone, and CHOP (cyclophosphamide,

83
Robak

hydroxyrubicin, vincristine and prednisone) and Autoimmune hemolytic anemia occurring after
achieved only short PR after each line of therapy. hematopoietic stem cell transplantation is poorly
Subsequently, he was treated with rituximab at a responsive to corticosteroids. The response to i.v.
dose of 375 mg/m2/wk for 4 wk. The first infusion of Ig or splenectomy is usually also unsatisfactory.
MoAb induced a significant reduction of lympho- However, recent reports of this complication with
cytes and after 5 d the hemoglobin (Hb) level started rituximab treatment seem to be encouraging (40–
to increase. At the end of week 8 there were no signs 42). Hongeng and colleagues observed a case of
of AIHA with normal values of reticulocytes and pediatric b-thalassemia major patients who
hemoglobin and only slightly positive direct anti- underwent unrelated hematopoietic stem cell trans-
globulin test (DAT) diagnosed as PR according to plantation and developed AIHA refractory to
NCI criteria lasting 2 months at the time of publi- corticosteroids and i.v. Ig therapy (42). Subse-
cation. Successful treatment with rituximab of ster- quently, he received rituximab 375 mg/m2 once
oid and cyclophosphamide-resistant hemolysis in a weekly at a total of two doses. Hemolysis was
CLL patient was also described by Chemnitz and noted to be decreased during 3 months of obser-
colleagues (35). The patient’s condition rapidly vation after treatment and the corticosteroid was
improved after standard doses of rituximab and the tapered off and discontinued in a month after the
hemolysis parameters tended to normalize on a second infusion of MoAb. Another child with
slightly elevated plateau. The steroid doses could be unrelated hematopoietic stem cell transplantation
reduced to 10 mg of prednisone per day. complicated by steroid refractory AIHA and
Combined use of rituximab with cytotoxic agents successfully treated with rituximab has been
can be even more effective in the treatment of recently presented by Corti and colleagues (40).
AIHA in the course of CLL. Gupta and colleagues She received three standard doses of rituximab
(33) treated eight patients with steroid refractory and Hb reached the level of 10 g/dL 15 d after the
AIHA in patients with CLL using rituximab at a first dose of MoAb. Direct Coombs test positivity
dose 375 mg/m2 i.v. on day 1, cyclophosphamide at progressively declined and she subsequently
a dose 750 mg/m2 on day 2 and dexamethasone at a become negative. Successful treatment of AIHA
dose 12 mg given intravenously on days 1 and 2 complicating haplo-identical hematopoietic stem
and orally from day 3 to 7. Treatment cycles were cell transplantation was reported by Ship et al.
repeated at 4-wk intervals till a best response was (41). A 7-yr-old patient received rituximab
achieved. All eight patients achieved remission of 375 mg/m2 weekly at four doses and hemoglobin
AIHA with a median hemoglobin level of 14.2 g/dL level stabilized at 11 g/dL 1 wk prior to the final
following treatment and a median duration of dose. He has not had a recurrence of his hemolytic
hemoglobin response of 13 months. The increase in anemia 1 yr after the treatment, at the time of
hemoglobin was evident as early as two cycles. publication. These three case reports indicate that
Moreover, five patients who were strongly Coombs rituximab is an active agent for the treatment of
positive before treatment turned to Coombs negat- AIHA complicating hematopoietic stem cell trans-
ive. It is worth noting that five patients who plantation and provide a basis for future pros-
relapsed following an initial response were pective trials.
re-treated with this regimen and all of them respon- Experience with the use of alemtuzumab in
ded with long-lasting remission. patients with AIHA is very limited. Willis and
Further evidence for efficacy of rituximab in the colleagues (4) treated four patients with
treatment of refractory AIHA comes from the alemtuzumab, administered at a dose of 10 mg/d
patients with SLE. Perotta and colleagues (43) as an i.v. infusion for 10 d. Complete remission was
observed an 18-yr-old girl with SLE who developed observed in one patient and PR in two. The
AIHA that did not respond to conventional treat- responding patients had warm-type AIHA. One
ment with steroids, azathioprine and cyclosporine. patient with cold-type AIHA had a partial response
Her hemolytic disorder markedly ameliorated after with reduction in red cell transfusion requirements.
treatment with rituximab, starting a few days after Unfortunately, one patient with strongly positive
therapy and the patient remained disease-free DAT did not respond to treatment with
7 months later, at the time of writing the paper. alemtuzumab and died of intractable hemolysis
A patient with AIHA associated with SLE was also and systemic venous thrombosis. These preliminary
among the six patients described by Quartier and results indicate that alemtuzumab can induce
colleagues (29). This 1-yr-old child also responded response in severe AIHA patients who failed to
to rituximab therapy. Red blood cell transfusions respond to conventional immunosuppression. Lar-
were discontinued 14 d after the start of rituximab ger and prospective trials of this MoAb in the
and normal hemoglobin concentration and reticu- treatment of AIHA who have failed first-line
locyte counts were achieved within 4 months. therapy with steroids are desirable.

84
Monoclonal antibodies in autoimmune cytopenias

improved during rituximab therapy. It is unlikely


Cold agglutinin disease
that these additional agents play a role in inducing
Cold agglutinin disease (CAD) is an uncommon remission of hemolysis but they may influence the
hemolytic anemia associated with B-cell lymphopro- underlying lymphoproliferative diseases.
liferative disorders. This rare occurrence is related to Significant improvement has also been shown in
the production of anti-erythrocyte Ig by neoplastic a patient with refractory mixed cryoglobulin syn-
cells. Corticosteroids are of little value in treating pri- drome (type 2) (56) and in a patient with mixed
mary CAD. Other therapeutic approaches include warm and cold AIHA not associated with lympho-
alkylating agents, interferon, cladribine and splen- proliferative disease who relapsed after tapering of
ectomy, but they are also usually not effective. corticosteroids (52).
Several small prospective studies and some case These preliminary results indicate that rituximab
reports have demonstrated the beneficial effects of may represent an effective option to conventional
rituximab on CAD associated with the clonal immunosuppressive therapy for the treatment of
lymphoproliferation as well as idiopathic disease CAD and other autoimmune hemolytic disorders.
(Table 3) (45–55). Among the 28 observations pub- However, further studies of this MoAb in patients
lished, the rate of response was 16 (89%) of which with this diseases are warranted.
eight (44%) achieved CR (Table 3). These results
suggest that rituximab may represent a true alter-
Monoclonal antibodies in the treatment of pure red cell
native to conventional immunosuppressive therapy
aplasia
for the treatment of CAD. Berentsen et al. (45) in a
prospective study treated six patients with clonal Pure red cell aplasia is a rare hematological syn-
CD20+ j+ B-cell lymphoproliferations with CAD. drome that results from an isolated depletion of
Three patients were previously untreated and three erythroid precursors from the bone marrow. The
pretreated with corticosteroids, chlorambucil, cyclo- disease may present in acute or chronic form,
phosphamide and/or cladribine. One patient frequently in association with thymomas, lympho-
achieved CR and three PR. Two patients failed to proliferative disorders, autoimmune diseases, treat-
show any response to rituximab despite high levels of ment with certain drugs and infections. Primary
CD20 expression on the neoplastic cells. Similar PRCA is considered to result from the presence of a
results, were observed by Camou and colleagues who soluble inhibitory factor which is localized to the Ig
treated five patients with four weekly rituximab fraction and targets undefined molecules on eryth-
infusions (53). The treatment a remission in all the roid cells at any stage of differentiation between
cases including four PR and one CR. Others single erythrocyte burst forming units (BFU-E) and eryth-
case reports presenting successful treatment of ster- roblasts. Treatment of PRCA usually involves ster-
oids and chemotherapy refractory CAD have also oids and/or immunosuppressive drugs including
been presented (46–49, 50, 54). cyclosporine and cyclophosphamide. However, only
In some patients, rituximab was used in combination about half of the patients respond to such therapy.
with IFN-a (47) or corticosteroids and cyclophosph- Recently, some case reports have suggested that
amide (50). However, in these patients hemolysis rituximab may effectively restore erythropoiesis in
Table 3. Studies evaluating efficacy of rituximab in cold agglutinin disease

Response
Number Age Patients' Response
Reference of patients (years) characteristic Treatment regimen OR (%) CR (%) duration Comments
2
Berentsen et al. (45) 6 55–80; Primary CAD, three R 375 mg/m /weekly · 4 4 (67) 1 (57) 6–14 m; One relapsed patient
mean 68 patients untreated median 11 m responded for R re-treatment
Camou et al. (53) 5 NR Primary CAD R 375 mg/m2/weekly · 4 5 (100) 1 (20) 9 and 11 m
Eneglhardt et al. (54) 2 50, 60 Refractory R 375 mg/m2/weekly · 4 2 (100) 2 (100)
Lee et al. (51) 1 75 Secondary NHL R 375 mg/m2/weekly · 4 1 1 NR Rapid and sustained response
Refractory to CY after R therapy
Layios et al. (47) 1 67 Secondary NHL R 375 mg/m2/weekly · 4 1 1 8m Patient was treated with IFN-a
Refractory simultaneously with R therapy
2
Zaja et al. (46) 1 72 Refractory to prednisone R 375 mg/m /weekly · 4 1 1 10 m
and AZ
Mori et al. (49) 1 52 Primary CAD R 375 mg/m2/weekly · 4 1 0 3+m Lymphoma cells disappeared
after R therapy
Cohen et al. (48) 1 65 Primary CAD R 375 mg/m2/weekly · 4 1 1 8+m
Total 18 50–80 16 (89) 8 (44) 3 € 14 m

CAD, cold agglutinin disease; R, rituximab; CY, cyclophosphamide; m, months; OR, overall response; CR, complete response; NHL, non-Hodkgin's lymphoma NR, not reported;
IFN-a, interferona; AZ, azathioprine.

85
Robak

Table 4. Studies evaluating rituximab in pure red cell aplasia

Reference Age Disease characteristic Previous treatment Treatment Response

Ghazal (59) 79 yr PRCA, CLL IVIG, P R 375 mg/m2/weekly · 8 CR of PRCA 10 + m


Ghazal (59) 47 yr PRCA, CLL FA R 375 mg/m2/weekly · 8 CR of PRCA 12 + m
Auner et al. (57) 68 yr Primary PRCA CS, ALG R 375 mg/m2/weekly · 3 CR of PRCA 12 + m
Zecca et al. (58) 18 m Acquired PRCA, AIHA MP, IVIG, CS R 375 mg/m2/weekly · 2 CR 5+m
Battle et al. (60) 58 yr PRCA, CLL P, Chl, CS R 375 mg/m2/weekly · 4 CR of PRCA 6+m
Total 18 m to 79 yr All refractory 100% CR

NHL, non-Hodgkin's lymphoma; PRCA, pure red cell aplasia; AIHA, autoimmune hemolytic anemia; CLL, chronic lymphocytic leukemia; CS, cyclosporine; MP, methyl
prednisolone; Chl, chlorambucil; IVIG, intravenous immunoglobulins; P, prednisone; R, rituximab; CR, complete response; FA, fludarabine; ALG, antilymphocyte globulin; yr, years;
m, months.

patients with acquired PRCA, even if immunosup- fludarabine treatment at a dose of 375 mg/m2/wk
pressive therapies have failed (57–63). The results are for 8 wk and her reticulocyte count increased
summarized in Table 4. Auner and colleagues trea- gradually from 0.1% to 10% by the eighth week
ted a 68-yr-old man with acquired PRCA refractory of rituximab therapy. A year after the initiation of
to conventional treatment including prednisone, rituximab treatment the patient’s hemoglobin and
erythropoietin and antithymocyte globulin (ATG) reticulocyte count were normal. Similar results were
with rituximab (57). Subsequently, he received presented by Battle and colleagues (59). They
rituximab at a dose of 375 mg/m2, administered reported a patient with CLL in prolymphocytic
weekly for a total of three doses. The rapid increase transformation who developed PRCA without
in reticulocyte counts a few days after first rituximab response to cyclosporin A but who responded to
infusion was observed. Simultaneously, almost rituximab therapy. MoAb was administered at
complete disappearance of CD19+ B cells from standard doses weekly for 4 wk. Six months after
the peripheral blood was seen. Three months after rituximab therapy the patient remained well with a
completion of therapy the reticulocyte count was stable hemoglobin level of 12.0 g/dL.
89 · 109/L and the hemoglobin had reached 12.8 g/ Pure red cell aplasia is also a well-recognized
dL without red cell transfusion. Similar observation complication of allogeneic hematopoietic cell trans-
was made by Zecca and colleagues who gave plantation. Its development poses difficult thera-
rituximab only twice in an 18-month-old girl (58). peutic problems. Maschan and colleagues reported
The treatment resulted in marked depletion of B cells successful treatment of PRCA with a single dose of
and rise in reticulocyte count and hemoglobin levels rituximab (200 mg/m2) in a child after major ABO-
leading to transfusion independence and normal incompatible peripheral blood allogeneic stem cell
values of these parameters for 5 months without transplantation for acquired aplastic anemia (61). It
therapy at the time of publication. This case report should be noted however, that a case of PRCA
suggests that lower doses of rituximab than those caused by chronic persistent parvovirus B19 infec-
needed for the treatment of lymphomas may be tion in a patient treated with rituximab for non-
highly effective in eliminating the erythropoietic Hodgkin’s lymphoma has been also reported (62).
inhibitor from the patient’s serum. Administration of i.v. Ig is an effective treatment of
Pure red cell aplasia occurs in approximately 5% this complication.
of CLL, most often in the course of disease but also Clinical experience with alemtuzumab in patients
at presentation. Two recent case reports suggest with PRCA is very limited. Nevertheless, Willis and
that rituximab is an effective treatment of this CLL colleagues treated four patients with PRCA with
complication (59, 60). Ghasal observed two alemtuzumab (4). Response was obtained in two
patients with B-cell CLL and PRCA, one detected patients at 2 months and at 6 wk, respectively, and
during de novo presentation and one during therapy two patients were non-responders. Recently,
with fludarabine (60). The first patient did not another patient with PRCA coexisting with CLL
respond to cyclosporin A, prednisone and i.v. Ig and unresponsive to corticosteroids and Ig was also
and then responded dramatically to rituximab successfully treated with alemtuzumab (63).
administered at 375 mg/m2/wk for eight consecu-
tive wk. His reticulocyte count rose to 25% and by
Conclusions
the eighth week on rituximab his hemoglobin level
was 12.1 g/dL and did not need a transfusion The results presented above indicate that MoAbs
during 8 months follow-up. The second patient are highly active and well tolerated in autoimmune
developed PRCA after five cycles of fludarabine. cytopenias. In particular, rituximab appeared to
Rituximab was administered 6 wk after last cycle of have significant efficacy in the treatment of refract-

86
Monoclonal antibodies in autoimmune cytopenias

ory or relapsed ITP, AIHA, CAD and PRCA, 12. British Committee for Standards in Haematology General
refractory or relapsed after conventional immuno- Haematology Task Force. Guidelines for the investigation
and management of idiopathic thrombocytopenic purpura
suppressive therapies. Alemtuzumab has not been in adults, children and in pregnancy. Br J Haematol
used very often in the treatment of these disorders. 2003;120:574–596.
However, very preliminary results in the single 13. Yang R, Han ZC. Pathogenesis and management of
centers may indicate that this drug is also poten- chronic idiopathic thrombocytopenic purpura: an update.
Int J Hematol 2000;71:18–24.
tially useful in autoimmune disorders. An expand- 14. Stasi R, Pagano A, Stipa E, Amadori S. Rituximab chimeric
ing group of MoAbs directed against antigens on anti-CD20 monoclonal antibody treatment for adults with
the cells of immune system provides great promise chronic thrombocytopenic purpura. Blood 2001;98:952–957.
for therapeutic advances in patients with autoim- 15. Aggarwal A, Catlett JP. Rituximab: an anti-CD20
antibody for the treatment of chronic refractory immune
mune cytopenias. However, it should be noted that thrombocytopenic purpura. South Med J 2002;95:1209–
MoAbs are still experimental therapies in these 1212.
diseases. The majority of available data come from 16. Stasi R, Stipa E, Forte V, Meo P, Amadori S. Variable
retrospective, uncontrolled studies or even single patterns of response to rituximab treatment in adults with
chronic idiopathic thrombocytopenia purpura. Blood
case reports. Currently prospective controlled stud- 2002;99:3872–3873.
ies are not available. In the future prospective 17. Faurschou M, Hasselbalch HC, Nielsen OJ. Sustained
randomized studies, the benefit of MoAbs should remission of platelet counts following monoclonal
be weighed against their potential risks. In such anti-CD20 antibody therapy in two cases of idiopathic
studies the optimal schedules of MoAbs adminis- autoimmune thrombocytopenia and neutropenia. Eur
J Haematol 2001;66:408–411.
tration and the duration of the clinical effect should 18. Saleh MN, Gutheil J, Moore M, et al. A pilot study of
also be evaluated. the anti-CD20 monoclonal antibody rituximab in patients
with refractory immune thrombocytopenic. Semin Oncol
2000;27(Suppl. 12):99–103.
Acknowledgements 19. Zaja F, Vianelli N, Sperotto A, et al. B-cell compartment
as the selective target for the treatment of immune
This work was supported in part by grant from the Medical
thrombocytopenias. Haematologica 2003;88:538–546.
University of Lodz, Poland no. 503–106–2 and by the Foun-
20. Patel K, Berman J, Ferber A, Caro J. Refractory auto-
dation for the development of Diagnostics and Therapy,
immune thrombocytopenic purpura treatment with ritux-
Warsaw, Poland. I thank Ms Krystyna Marszalek for secre-
imab. Am J Hematol 2001;67:59–60.
tariat assistance.
21. Lim SH, Hale G, Marcus RE, Waldmann H, Baglin TP.
CAMPATH-1 monoclonal antibody therapy in severe
refractory autoimmune thrombocytopenic purpura. Br J
References Haematol 1993;84:542–544.
1. Marsh JC, Gordon-Smith EC. CAMPATH-1H in the 22. Cooper N, Bussel J. Rituximab treatment in patients with
treatment of autoimmune cytopenias. Cytotherapy immune thrombocytopenia purpura (ITP). Blood
2001;3:189–195. 2001;98:2180a (abstract).
2. Perotta A, Sunneberg TA, Scott J, et al. Rituximab in 23. Giagounidis AAN, Anhuf J, Schneider P, et al. Treat-
the treatment of chronic idiopathic thrombocytopenic ment of relapsed idiopathic thrombocytopenic purpura
purpura (ITP). Blood 1999;94:14a (abstract). with the anti-CD20 antibody rituximab. Blood
3. Silverman GJ, Weisman S. Rituximab therapy and auto- 2001;98:2187a (abstract).
immune disorders prospects for anti-B cell therapy. Arth- 24. Introna M, Golay J, Barbui T. Rituximab: a new
ritis Rheum 2003;48:1484–1492. therapeutic tool for primary immune thrombocytopenic
4. Willis F, Marsh JC, Bevan DH, et al. The effect of purpura. Haematologica 2003;88:482–484.
treatment with Campath-1H in patients with autoimmune 25. Hegde UP, Wilson WH, White T, Cheson BD. Ritux-
cytopenias. Br J Haematol 2001;114:891–898. imab treatment of refractory fludarabine-associated
5. Zaja F, De Vita S, Vianelli N, Ferraccioli G, Bacca- immune thrombocytopenia in chronic lymphocytic leuke-
rani M, Fanin R. The new therapeutic indications for mia. Blood 2002;100:2260–2262.
rituximab. Haematologica 2002; 88 (Suppl. 10): 20–22. 26. Ratanatharathorn V, Carson E, Reynolds C, et al.
6. Giagounidis AA, Anhuf J, Schneider P, et al. Treatment Anti-CD20 chimeric monoclonal antibody treatment of
of relapsed idiopathic thrombocytopenic purpura with the refractory immune-mediated thrombocytopenia in a patient
anti-CD20 monoclonal antibody rituximab: a pilot study. with chronic graft versus host disease. Ann Intern Med
Eur J Haematol 2002;69:95–100. 2000;133:275–279.
7. Zecca M, Nobili B, Ramenghi U, et al. Rituximab for the 27. Hensel M, Ho AD. Successful treatment of a patient with
treatment of refractory autoimmune hemolytic anemia in hairy cell leukemia and pentostatin induced autoimmune
children. Blood 2003;101:3857–3861. thrombocytopenia with rituximab. Am J Haematol
8. Plosker GL, Figgitt DP. Rituximab: a review of its use in 2003;73:37–40.
non-Hodgkin’s lymphoma and chronic lymphocytic leuk- 28. Thomas AT. Autoimmune hemolytic anemias. In: Wint-
aemia. Drugs 2003;63:803–843. robe S, Lee GR, Foerster J, Lukens J, Paraskevas F,
9. Frampton JE, Wagstaff AJ. Alemtuzumab. Drugs Greer JP, Rodgers GM, eds. Clinical Haematology, 10th
2003;63:1229–1243. edn. Philadelphia, PA: Lippincott Williams and Wilkins,
10. Robak T. Monoclonal antibodies in the treatment of chronic 1998:1233–1263.
lymphoid leukemias. Leuk Lymphoma 2004;45:205–219. 29. Quartier P, Brethon B, Philippet P, Landman-Parker
11. Treon SP, Anderson KC. The use of rituximab in the J, Le Deist F, Fischer A. Treatment of childhood auto-
treatment of malignant and nonmalignant plasma cell dis- immune haemolytic anaemia with rituximab. Lancet
orders. Semin Oncol 2000;27:79–85. 2001;358:1511–1513.

87
Robak

30. Lee E, Zamkoff KW, Gentile TC, Zimrin A. Rituxam in 47. Lavios N, Van Den Neste E, Jost E, Deneys V, Scheiff
the treatment of autoimmune haemolytic anemia (AIHA). JM, Ferrant A. Remission of severe cold agglutinin
Blood 2000;96:2560 (abstract). disease after rituximab therapy. Leukemia 2001;15:187–
31. Reddy S, Schwartz JM, Hussain S, Fernandez G. Rit- 188.
uximab-based therapy in 3 patients with autoimmune 48. Cohen Y, Polliack A, Zelig O, Goldfarb A. Mono-
hemolytic anemia. Blood 2002;98:3610 (abstract). therapy with rituximab induces rapid remission of recurrent
32. Ahrens N, Kingreen D, Seltsam A, Salama A. Treat- cold agglutinin-mediated hemolytic anemia in a patient
ment of refractory autoimmune haemolytic anaemia with with indolent lympho-plasmacytic lymphoma. Leuk Lym-
anti-CD20 (rituximab). Br J Hematol 2001;114:244–245. phoma 2001;42:1405–1408.
33. Gupta N, Kuvaru S, Patel D, et al. Rituximab based 49. Mori A, Tamaru J, Sumi H, Kondo H. Beneficial effects of
chemotherapy for steroid-refractory autoimmune hemo- rituximab on primary cold agglutinin disease refractory to
lytic anemia of chronic lymphocytic leukemia. Leukemia conventional therapy. Eur J Haematol 2002;68:243–246.
2002;16:2092–2095. 50. Bauduer F. Rituximab: a very efficient therapy in cold
34. Seipelt G, Bohme A, Koschmieder S, Hoelzer D. agglutinins and refractory autoimmune haemolytic anae-
Effective treatment with rituximab in a patient with mia associated with CD20 positive, low-grade non Hodg-
refractory prolymphocytoid transformed B-chronic kin’s lymphoma. Br J Haematol 2001;112:1085–1086.
lymphocytic leukemia and Evans syndrome. Ann Hematol 51. Lee EJ, Kueck B. Rituximab in the treatment of cold
2001;80:170–173. agglutinin disease. Blood 1998;92:3490–3491.
35. Chemnitz J, Draube A, Diehl V, Wolf J. Successful 52. Morselli M, Luppi M, Potenza L, et al. Mixed warm and
treatment of steroid and cyclophosphamide-resistant chro- cold autoimmune hemolytic anemia: complete recovery
nic lymphocytic leukemia with rituximab. Am J Hematol after 2 courses of rituximab treatment. Blood
2002;69:232–233. 2002;99:3478–3479.
36. Trape G, Fianchi L, Lai M, et al. Rituximab chimeric 53. Camou F, Viallard IF, Pellegrin JL. Rituximab in cold
anti-CD20 monoclonal antibody treatment for refractory agglutinin disease. Rev Med Interne 2003;24:501–504.
hemolytic anemia in patients with lymphoproliferative 54. Engelhardt M, Jakob A, Ruter B, Trepel M, Hirsch F,
disorders. Haematologica 2003;88:223–225. Lubbert M. Severe cold hemaglutinin disease (CHD) suc-
37. Iannitto E, Ammatuna E, Marino C, Cirrincione S, cessfully treated with rituximab. Blood 2002;100:1922–
Greco G, Mariani G. Sustained response of refractory 1923.
chronic lymphocytic leukemia in progression complicated 55. Sparling TG, Andricevic M, Wass H. Remission of cold
by acute hemolitic anemia to anti-CD20 monoclonal anti- hemagglutinin disease induced by rituximab therapy.
body. Blood 2002;99:1096–1097. CMAJ 2001;164:1405.
38. Motto DG, Williams JA, Boxer LA. Rituximab for 56. Zaja F, Russo D, Fuga G, Patriarca F, Ermacora A,
refractory childhood autoimmune hemolytic anemia. Isr Baccarani M. Rituximab for the treatment of type II
Med Assoc J 2002;4:1006–1008. mixed cryoglobulinemia. Haematologica 1999;84:1157–
39. Wong S, McKendrick J. Rituximab in lymphoma with life 1158.
threatening haemolysis. Leuk Lymphoma 2002;43:1711– 57. Auner HW, Wolfler A, Beham-Schmid C, Strunk D,
1712. Linkesch W, Sill H. Restoration of erythropoiesis by
40. Corti P, Bonanomi S, Vallinoto C, et al. Rituximab for rituximab in an adult patient with primary acquired pure
immune hemolytic anemia following T and B-cell depleted red cell aplasia refractory to conventional treatment. Br J
hematopoietic stem cell transplantation. Acta Haematol Haematol 2002;116:727–728.
2003;109:43–45. 58. Zecca M, DeStefano P, Nobili B, Locatelli F. Anti-
41. Ship A, May W, Lucas K. Anti-CD20 monoclonal anti- CD20 monoclonal antibody for the treatment of severe
body therapy for autoimmune hemolytic anemia following immuno-mediated, pure red cell aplasia and hemolytic
T cell depleted haploidentical stem cell transplantation. anemia. Blood 2001;97:3995–3997.
Bone Marrow Trans 2002;29:365–366. 59. Ghazal H. Successful treatment of pure red cell aplasia
42. Hongeng S, Tardtong P, Worapongpaiboon S, Ungk- with rituximab in patients with chronic lymphocytic le-
anont A, Jootar S. Successful treatment of refractory ukemia. Blood 2002;99:1092–1094.
autoimmune haemolytic anaemia in a post unrelated bone 60. Batlle M, Ribera JM, Oriol A, Plensa E, Milla F,
marrow transplant pediatric patient with rituximab. Bone Felin E. Successful response to rituximab in a patient with
Marrow Trans 2002;29:871–872. pure red cell aplasia complicating chronic lymphocytic
43. Perotta S, Locatelli F, La Manna A, Cennamo L, leukemia. Br J Haematol 2002;118:1192–1193.
De Stefano P, Nobili B. Anti-CD20 monoclonal antibody 61. Maschan AA, Skorobogatova EV, Balashow DN, et al.
(rituximab) for life-threatening autoimmune haemolytic Successful treatment of pure red cell aplasia with a single
anaemia in a patient with systemic lupus erythematosus. Br dose of rituximab in a child after major AB incompatible
J Haematol 2002;116:465–467. peripheral blood allogenic stem cell transplantation for
44. Mc Mahon C, Babu L, Hodgson A, Hayat A, Connell acquired aplastic anemia. Bone Marrow Transpl
NO, Smith OP. Childhood refractory autoimmune hae- 2002;30:405–407.
molytic anaemia: is there a role for anti-CD20 therapy 62. Sharma VR, Fleming DR, Slone SP. Pure red cell aplasia
(rituximab). Br J Haematol 2002;117:480–483. due to parvovirus B19 in a patient treated with rituximab.
45. Berentsen S, Tjonnfjord GE, Brudevold R, et al. Blood 2000;96:1184–1186.
Favourable response to therapy with the anti-CD20 63. Rodon P, Breton P, Courouble G. Treatment of pure red
monoclonal antibody rituximab in primary chronic cold cell aplasia and autoimmune haemolytic anaemia in chronic
agglutinin disease. Br J Haematol 2001;115:79–83. lymphocytic leukemia with Campath-1H. Eur J Haematol
46. Zaja F, Russo D, Fuga G, et al. Rituximab in a case of 2003;70:319–321.
cold agglutinin disease. Br J Haematol 2001;115:
230–233.

88

You might also like